Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 177.603
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Annu Rev Immunol ; 40: 589-614, 2022 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-35130029

RESUMO

Pulmonary granulomas are widely considered the epicenters of the immune response to Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB). Recent animal studies have revealed factors that either promote or restrict TB immunity within granulomas. These models, however, typically ignore the impact of preexisting immunity on cellular organization and function, an important consideration because most TB probably occurs through reinfection of previously exposed individuals. Human postmortem research from the pre-antibiotic era showed that infections in Mtb-naïve individuals (primary TB) versus those with prior Mtb exposure (postprimary TB) have distinct pathologic features. We review recent animal findings in TB granuloma biology, which largely reflect primary TB. We also discuss our current understanding of postprimary TB lesions, about which much less is known. Many knowledge gaps remain, particularly regarding how preexisting immunity shapes granuloma structure and local immune responses at Mtb infection sites.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Animais , Granuloma/etiologia , Humanos , Pulmão/microbiologia , Pulmão/patologia
2.
Annu Rev Immunol ; 39: 611-637, 2021 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-33637017

RESUMO

Infection with Mycobacterium tuberculosis causes >1.5 million deaths worldwide annually. Innate immune cells are the first to encounter M. tuberculosis, and their response dictates the course of infection. Dendritic cells (DCs) activate the adaptive response and determine its characteristics. Macrophages are responsible both for exerting cell-intrinsic antimicrobial control and for initiating and maintaining inflammation. The inflammatory response to M. tuberculosis infection is a double-edged sword. While cytokines such as TNF-α and IL-1 are important for protection, either excessive or insufficient cytokine production results in progressive disease. Furthermore, neutrophils-cells normally associated with control of bacterial infection-are emerging as key drivers of a hyperinflammatory response that results in host mortality. The roles of other innate cells, including natural killer cells and innate-like T cells, remain enigmatic. Understanding the nuances of both cell-intrinsic control of infection and regulation of inflammation will be crucial for the successful development of host-targeted therapeutics and vaccines.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Animais , Citocinas , Humanos , Imunidade Inata , Macrófagos
3.
Annu Rev Immunol ; 36: 603-638, 2018 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-29490165

RESUMO

Globally, about 36.7 million people were living with HIV infection at the end of 2015. The most frequent infection co-occurring with HIV-1 is Mycobacterium tuberculosis-374,000 deaths per annum are attributable to HIV-tuberculosis, 75% of those occurring in Africa. HIV-1 infection increases the risk of tuberculosis by a factor of up to 26 and alters its clinical presentation, complicates diagnosis and treatment, and worsens outcome. Although HIV-1-induced depletion of CD4+ T cells underlies all these effects, more widespread immune deficits also contribute to susceptibility and pathogenesis. These defects present a challenge to understand and ameliorate, but also an opportunity to learn and optimize mechanisms that normally protect people against tuberculosis. The most effective means to prevent and ameliorate tuberculosis in HIV-1-infected people is antiretroviral therapy, but this may be complicated by pathological immune deterioration that in turn requires more effective host-directed anti-inflammatory therapies to be derived.


Assuntos
Coinfecção , Infecções por HIV/imunologia , HIV-1/imunologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Animais , Terapia Antirretroviral de Alta Atividade , Antituberculosos/farmacologia , Antituberculosos/uso terapêutico , Progressão da Doença , Variação Genética , Infecções por HIV/diagnóstico , Infecções por HIV/terapia , Infecções por HIV/virologia , HIV-1/genética , Humanos , Tuberculose/diagnóstico , Tuberculose/microbiologia , Tuberculose/terapia , Replicação Viral
4.
Cell ; 186(23): 5135-5150.e28, 2023 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-37865090

RESUMO

Mycobacterium tuberculosis (Mtb) cultured axenically without detergent forms biofilm-like cords, a clinical identifier of virulence. In lung-on-chip (LoC) and mouse models, cords in alveolar cells contribute to suppression of innate immune signaling via nuclear compression. Thereafter, extracellular cords cause contact-dependent phagocyte death but grow intercellularly between epithelial cells. The absence of these mechanopathological mechanisms explains the greater proportion of alveolar lesions with increased immune infiltration and dissemination defects in cording-deficient Mtb infections. Compression of Mtb lipid monolayers induces a phase transition that enables mechanical energy storage. Agent-based simulations demonstrate that the increased energy storage capacity is sufficient for the formation of cords that maintain structural integrity despite mechanical perturbation. Bacteria in cords remain translationally active despite antibiotic exposure and regrow rapidly upon cessation of treatment. This study provides a conceptual framework for the biophysics and function in tuberculosis infection and therapy of cord architectures independent of mechanisms ascribed to single bacteria.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Animais , Camundongos , Biofilmes , Pulmão/microbiologia , Pulmão/patologia , Mycobacterium tuberculosis/fisiologia , Tuberculose/microbiologia , Tuberculose/patologia , Virulência , Fenômenos Biomecânicos
5.
Cell ; 186(23): 4994-4995, 2023 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-37949055

RESUMO

Mechanobiology explores how cells sense and respond to mechanical cues and how mechanics guide cell function, physiology, and disease. In this issue of Cell, Thacker and colleagues reveal how the tuberculosis-causing pathogen exploits the mechanical behavior of cord-like structures to promote infection, impacting immune response, antibiotic susceptibility, and treatment strategies.


Assuntos
Fenômenos Biomecânicos , Mycobacterium tuberculosis , Humanos , Biofísica , Tuberculose/microbiologia , Mycobacterium tuberculosis/fisiologia
6.
Cell ; 186(5): 1013-1025.e24, 2023 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-36827973

RESUMO

The emergence of drug-resistant tuberculosis has created an urgent need for new anti-tubercular agents. Here, we report the discovery of a series of macrolides called sequanamycins with outstanding in vitro and in vivo activity against Mycobacterium tuberculosis (Mtb). Sequanamycins are bacterial ribosome inhibitors that interact with the ribosome in a similar manner to classic macrolides like erythromycin and clarithromycin, but with binding characteristics that allow them to overcome the inherent macrolide resistance of Mtb. Structures of the ribosome with bound inhibitors were used to optimize sequanamycin to produce the advanced lead compound SEQ-9. SEQ-9 was efficacious in mouse models of acute and chronic TB as a single agent, and it demonstrated bactericidal activity in a murine TB infection model in combination with other TB drugs. These results support further investigation of this series as TB clinical candidates, with the potential for use in new regimens against drug-susceptible and drug-resistant TB.


Assuntos
Antituberculosos , Mycobacterium tuberculosis , Animais , Camundongos , Antituberculosos/farmacologia , Macrolídeos , Farmacorresistência Bacteriana , Claritromicina
7.
Cell ; 186(10): 2176-2192.e22, 2023 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-37137307

RESUMO

The ClpC1:ClpP1P2 protease is a core component of the proteostasis system in mycobacteria. To improve the efficacy of antitubercular agents targeting the Clp protease, we characterized the mechanism of the antibiotics cyclomarin A and ecumicin. Quantitative proteomics revealed that the antibiotics cause massive proteome imbalances, including upregulation of two unannotated yet conserved stress response factors, ClpC2 and ClpC3. These proteins likely protect the Clp protease from excessive amounts of misfolded proteins or from cyclomarin A, which we show to mimic damaged proteins. To overcome the Clp security system, we developed a BacPROTAC that induces degradation of ClpC1 together with its ClpC2 caretaker. The dual Clp degrader, built from linked cyclomarin A heads, was highly efficient in killing pathogenic Mycobacterium tuberculosis, with >100-fold increased potency over the parent antibiotic. Together, our data reveal Clp scavenger proteins as important proteostasis safeguards and highlight the potential of BacPROTACs as future antibiotics.


Assuntos
Antituberculosos , Mycobacterium tuberculosis , Antituberculosos/farmacologia , Proteínas de Bactérias/metabolismo , Endopeptidase Clp/metabolismo , Proteínas de Choque Térmico/metabolismo , Mycobacterium tuberculosis/efeitos dos fármacos , Proteostase
8.
Cell ; 186(25): 5536-5553.e22, 2023 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-38029747

RESUMO

Mycobacterium tuberculosis (Mtb) causes 1.6 million deaths annually. Active tuberculosis correlates with a neutrophil-driven type I interferon (IFN) signature, but the cellular mechanisms underlying tuberculosis pathogenesis remain poorly understood. We found that interstitial macrophages (IMs) and plasmacytoid dendritic cells (pDCs) are dominant producers of type I IFN during Mtb infection in mice and non-human primates, and pDCs localize near human Mtb granulomas. Depletion of pDCs reduces Mtb burdens, implicating pDCs in tuberculosis pathogenesis. During IFN-driven disease, we observe abundant DNA-containing neutrophil extracellular traps (NETs) described to activate pDCs. Cell-type-specific disruption of the type I IFN receptor suggests that IFNs act on IMs to inhibit Mtb control. Single-cell RNA sequencing (scRNA-seq) indicates that type I IFN-responsive cells are defective in their response to IFNγ, a cytokine critical for Mtb control. We propose that pDC-derived type I IFNs act on IMs to permit bacterial replication, driving further neutrophil recruitment and active tuberculosis disease.


Assuntos
Interferon Tipo I , Tuberculose , Humanos , Camundongos , Animais , Macrófagos/microbiologia , Citocinas , Neutrófilos , Células Dendríticas
9.
Cell ; 185(25): 4682-4702, 2022 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-36493751

RESUMO

Despite having been identified as the organism that causes tuberculosis in 1882, Mycobacterium tuberculosis has managed to still evade our understanding of the protective immune response against it, defying the development of an effective vaccine. Technology and novel experimental models have revealed much new knowledge, particularly with respect to the heterogeneity of the bacillus and the host response. This review focuses on certain immunological elements that have recently yielded exciting data and highlights the importance of taking a holistic approach to understanding the interaction of M. tuberculosis with the many host cells that contribute to the development of protective immunity.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Humanos , Comunicação Celular , Tuberculose/imunologia
10.
Cell ; 185(24): 4507-4525.e18, 2022 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-36356582

RESUMO

The human pathogen Mycobacterium tuberculosis typically causes lung disease but can also disseminate to other tissues. We identified a M. tuberculosis (Mtb) outbreak presenting with unusually high rates of extrapulmonary dissemination and bone disease. We found that the causal strain carried an ancestral full-length version of the type VII-secreted effector EsxM rather than the truncated version present in other modern Mtb lineages. The ancestral EsxM variant exacerbated dissemination through enhancement of macrophage motility, increased egress of macrophages from established granulomas, and alterations in macrophage actin dynamics. Reconstitution of the ancestral version of EsxM in an attenuated modern strain of Mtb altered the migratory mode of infected macrophages, enhancing their motility. In a zebrafish model, full-length EsxM promoted bone disease. The presence of a derived nonsense variant in EsxM throughout the major Mtb lineages 2, 3, and 4 is consistent with a role for EsxM in regulating the extent of dissemination.


Assuntos
Doenças Ósseas , Mycobacterium marinum , Mycobacterium tuberculosis , Tuberculose , Animais , Humanos , Peixe-Zebra , Tuberculose/microbiologia , Macrófagos/microbiologia , Proteínas de Bactérias/genética
11.
Cell ; 185(24): 4467-4469, 2022 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-36423577

RESUMO

In this issue of Cell, Saelens et al. describe a new function for mycobacterial Type VII secretion systems: manipulation of host cell migration. They find that a substantial proportion of global TB cases arise from bacteria lacking this function, raising questions about its role in pathoadaptation of Mycobacterium tuberculosis.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Sistemas de Secreção Tipo VII , Humanos , Sistemas de Secreção Tipo VII/metabolismo , Mycobacterium tuberculosis/metabolismo , Tuberculose/microbiologia , Movimento Celular
12.
Cell ; 185(20): 3720-3738.e13, 2022 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-36103894

RESUMO

Necrosis of macrophages in the granuloma, the hallmark immunological structure of tuberculosis, is a major pathogenic event that increases host susceptibility. Through a zebrafish forward genetic screen, we identified the mTOR kinase, a master regulator of metabolism, as an early host resistance factor in tuberculosis. We found that mTOR complex 1 protects macrophages from mycobacterium-induced death by enabling infection-induced increases in mitochondrial energy metabolism fueled by glycolysis. These metabolic adaptations are required to prevent mitochondrial damage and death caused by the secreted mycobacterial virulence determinant ESAT-6. Thus, the host can effectively counter this early critical mycobacterial virulence mechanism simply by regulating energy metabolism, thereby allowing pathogen-specific immune mechanisms time to develop. Our findings may explain why Mycobacterium tuberculosis, albeit humanity's most lethal pathogen, is successful in only a minority of infected individuals.


Assuntos
Mycobacterium marinum , Mycobacterium tuberculosis , Tuberculose , Animais , Mycobacterium tuberculosis/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Peixe-Zebra
13.
Cell ; 185(17): 3086-3103, 2022 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-35985287

RESUMO

The immense interindividual clinical variability during any infection is a long-standing enigma. Inborn errors of IFN-γ and IFN-α/ß immunity underlying rare infections with weakly virulent mycobacteria and seasonal influenza virus have inspired studies of two common infections: tuberculosis and COVID-19. A TYK2 genotype impairing IFN-γ production accounts for about 1% of tuberculosis cases, and autoantibodies neutralizing IFN-α/ß account for about 15% of critical COVID-19 cases. The discovery of inborn errors and mechanisms underlying rare infections drove the identification of common monogenic or autoimmune determinants of related common infections. This "rare-to-common" genetic and mechanistic approach to infectious diseases may be of heuristic value.


Assuntos
COVID-19 , Mycobacterium , Tuberculose , Humanos , Interferon-alfa , Interferon beta , Interferon gama
14.
Nat Immunol ; 25(8): 1411-1421, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38997431

RESUMO

A subset of individuals exposed to Mycobacterium tuberculosis (Mtb) that we refer to as 'resisters' (RSTR) show evidence of IFN-γ- T cell responses to Mtb-specific antigens despite serially negative results on clinical testing. Here we found that Mtb-specific T cells in RSTR were clonally expanded, confirming the priming of adaptive immune responses following Mtb exposure. RSTR CD4+ T cells showed enrichment of TH17 and regulatory T cell-like functional programs compared to Mtb-specific T cells from individuals with latent Mtb infection. Using public datasets, we showed that these TH17 cell-like functional programs were associated with lack of progression to active tuberculosis among South African adolescents with latent Mtb infection and with bacterial control in nonhuman primates. Our findings suggested that RSTR may successfully control Mtb following exposure and immune priming and established a set of T cell biomarkers to facilitate further study of this clinical phenotype.


Assuntos
Linfócitos T CD4-Positivos , Mycobacterium tuberculosis , Tuberculose , Mycobacterium tuberculosis/imunologia , Humanos , Animais , Adolescente , Tuberculose/imunologia , Tuberculose/microbiologia , Linfócitos T CD4-Positivos/imunologia , Células Th17/imunologia , Feminino , Macaca mulatta , Masculino , Fenótipo , Interferon gama/metabolismo , Interferon gama/imunologia , Antígenos de Bactérias/imunologia , Tuberculose Latente/imunologia , Tuberculose Latente/microbiologia , África do Sul , Adulto Jovem , Linfócitos T Reguladores/imunologia , Adulto
15.
Cell ; 184(17): 4579-4592.e24, 2021 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-34297925

RESUMO

Antibacterial agents target the products of essential genes but rarely achieve complete target inhibition. Thus, the all-or-none definition of essentiality afforded by traditional genetic approaches fails to discern the most attractive bacterial targets: those whose incomplete inhibition results in major fitness costs. In contrast, gene "vulnerability" is a continuous, quantifiable trait that relates the magnitude of gene inhibition to the effect on bacterial fitness. We developed a CRISPR interference-based functional genomics method to systematically titrate gene expression in Mycobacterium tuberculosis (Mtb) and monitor fitness outcomes. We identified highly vulnerable genes in various processes, including novel targets unexplored for drug discovery. Equally important, we identified invulnerable essential genes, potentially explaining failed drug discovery efforts. Comparison of vulnerability between the reference and a hypervirulent Mtb isolate revealed incomplete conservation of vulnerability and that differential vulnerability can predict differential antibacterial susceptibility. Our results quantitatively redefine essential bacterial processes and identify high-value targets for drug development.


Assuntos
Regulação Bacteriana da Expressão Gênica , Genoma Bacteriano , Mycobacterium tuberculosis/genética , Aminoacil-tRNA Sintetases/metabolismo , Antituberculosos/farmacologia , Teorema de Bayes , Evolução Biológica , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Mycobacterium tuberculosis/efeitos dos fármacos , RNA Guia de Cinetoplastídeos/genética
16.
Nat Immunol ; 24(5): 855-868, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37012543

RESUMO

Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is a global cause of death. Granuloma-associated lymphoid tissue (GrALT) correlates with protection during TB, but the mechanisms of protection are not understood. During TB, the transcription factor IRF4 in T cells but not B cells is required for the generation of the TH1 and TH17 subsets of helper T cells and follicular helper T (TFH)-like cellular responses. A population of IRF4+ T cells coexpress the transcription factor BCL6 during Mtb infection, and deletion of Bcl6 (Bcl6fl/fl) in CD4+ T cells (CD4cre) resulted in reduction of TFH-like cells, impaired localization within GrALT and increased Mtb burden. In contrast, the absence of germinal center B cells, MHC class II expression on B cells, antibody-producing plasma cells or interleukin-10-expressing B cells, did not increase Mtb susceptibility. Indeed, antigen-specific B cells enhance cytokine production and strategically localize TFH-like cells within GrALT via interactions between programmed cell death 1 (PD-1) and its ligand PD-L1 and mediate Mtb control in both mice and macaques.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Camundongos , Animais , Linfócitos T Auxiliares-Indutores , Linfócitos B , Tecido Linfoide , Centro Germinativo , Fatores de Transcrição
17.
Annu Rev Immunol ; 31: 475-527, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23516984

RESUMO

There are 9 million cases of active tuberculosis reported annually; however, an estimated one-third of the world's population is infected with Mycobacterium tuberculosis and remains asymptomatic. Of these latent individuals, only 5-10% will develop active tuberculosis disease in their lifetime. CD4(+) T cells, as well as the cytokines IL-12, IFN-γ, and TNF, are critical in the control of Mycobacterium tuberculosis infection, but the host factors that determine why some individuals are protected from infection while others go on to develop disease are unclear. Genetic factors of the host and of the pathogen itself may be associated with an increased risk of patients developing active tuberculosis. This review aims to summarize what we know about the immune response in tuberculosis, in human disease, and in a range of experimental models, all of which are essential to advancing our mechanistic knowledge base of the host-pathogen interactions that influence disease outcome.


Assuntos
Tuberculose Pulmonar/imunologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Predisposição Genética para Doença , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Mycobacterium tuberculosis/imunologia , Resultado do Tratamento , Tuberculose Pulmonar/diagnóstico , Tuberculose Pulmonar/epidemiologia
18.
Annu Rev Immunol ; 31: 605-33, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23516986

RESUMO

Evidence has increasingly shown that the lungs are a major site of immune regulation. A robust and highly regulated immune response in the lung protects the host from pathogen infection, whereas an inefficient or deleterious response can lead to various pulmonary diseases. Many cell types, such as epithelial cells, dendritic cells, macrophages, neutrophils, eosinophils, and B and T lymphocytes, contribute to lung immunity. This review focuses on the recent advances in understanding how T lymphocytes mediate pulmonary host defenses against bacterial, viral, and fungal pathogens.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Pulmão/imunologia , Pulmão/patologia , Tuberculose Pulmonar/imunologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Pulmão/microbiologia , Linfonodos/imunologia , Linfonodos/microbiologia , Linfonodos/patologia , Mycobacterium tuberculosis/imunologia , Tuberculose Pulmonar/microbiologia , Tuberculose Pulmonar/patologia
19.
Cell ; 183(3): 752-770.e22, 2020 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-33125891

RESUMO

A greater understanding of hematopoietic stem cell (HSC) regulation is required for dissecting protective versus detrimental immunity to pathogens that cause chronic infections such as Mycobacterium tuberculosis (Mtb). We have shown that systemic administration of Bacille Calmette-Guérin (BCG) or ß-glucan reprograms HSCs in the bone marrow (BM) via a type II interferon (IFN-II) or interleukin-1 (IL1) response, respectively, which confers protective trained immunity against Mtb. Here, we demonstrate that, unlike BCG or ß-glucan, Mtb reprograms HSCs via an IFN-I response that suppresses myelopoiesis and impairs development of protective trained immunity to Mtb. Mechanistically, IFN-I signaling dysregulates iron metabolism, depolarizes mitochondrial membrane potential, and induces cell death specifically in myeloid progenitors. Additionally, activation of the IFN-I/iron axis in HSCs impairs trained immunity to Mtb infection. These results identify an unanticipated immune evasion strategy of Mtb in the BM that controls the magnitude and intrinsic anti-microbial capacity of innate immunity to infection.


Assuntos
Células-Tronco Hematopoéticas/microbiologia , Imunidade , Mycobacterium tuberculosis/fisiologia , Mielopoese , Animais , Células da Medula Óssea/metabolismo , Proliferação de Células , Suscetibilidade a Doenças , Homeostase , Interferon Tipo I/metabolismo , Ferro/metabolismo , Cinética , Pulmão/microbiologia , Pulmão/patologia , Macrófagos/imunologia , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Necrose , Transdução de Sinais , Transcrição Gênica , Tuberculose/imunologia , Tuberculose/microbiologia , Tuberculose/patologia
20.
Cell ; 181(2): 293-305.e11, 2020 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-32142653

RESUMO

Pulmonary tuberculosis, a disease caused by Mycobacterium tuberculosis (Mtb), manifests with a persistent cough as both a primary symptom and mechanism of transmission. The cough reflex can be triggered by nociceptive neurons innervating the lungs, and some bacteria produce neuron-targeting molecules. However, how pulmonary Mtb infection causes cough remains undefined, and whether Mtb produces a neuron-activating, cough-inducing molecule is unknown. Here, we show that an Mtb organic extract activates nociceptive neurons in vitro and identify the Mtb glycolipid sulfolipid-1 (SL-1) as the nociceptive molecule. Mtb organic extracts from mutants lacking SL-1 synthesis cannot activate neurons in vitro or induce cough in a guinea pig model. Finally, Mtb-infected guinea pigs cough in a manner dependent on SL-1 synthesis. Thus, we demonstrate a heretofore unknown molecular mechanism for cough induction by a virulent human pathogen via its production of a complex lipid.


Assuntos
Tosse/fisiopatologia , Glicolipídeos/metabolismo , Nociceptores/fisiologia , Fatores de Virulência/metabolismo , Adulto , Animais , Linhagem Celular , Tosse/etiologia , Tosse/microbiologia , Feminino , Glicolipídeos/fisiologia , Cobaias , Interações Hospedeiro-Patógeno , Humanos , Lipídeos/fisiologia , Pulmão/microbiologia , Macrófagos/microbiologia , Masculino , Camundongos , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidade , Cultura Primária de Células , Tuberculose/microbiologia , Tuberculose Pulmonar/microbiologia , Tuberculose Pulmonar/fisiopatologia , Fatores de Virulência/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA